Pan-HDAC Inhibitors Promote Tau Aggregation by Increasing the Level of Acetylated Tau

Total Page:16

File Type:pdf, Size:1020Kb

Pan-HDAC Inhibitors Promote Tau Aggregation by Increasing the Level of Acetylated Tau International Journal of Molecular Sciences Article Pan-HDAC Inhibitors Promote Tau Aggregation by Increasing the Level of Acetylated Tau 1,2, 1,3, 3,4 5 2 Hyeanjeong Jeong y, Seulgi Shin y, Jun-Seok Lee , Soo Hyun Lee , Ja-Hyun Baik , Sungsu Lim 1,* and Yun Kyung Kim 1,3,* 1 Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea 2 Department of Life Sciences, Korea University, Seoul 02841, Korea 3 Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon 34113, Korea 4 Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea 5 Center for Biomicrosystems, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea * Correspondence: [email protected] (S.L.); [email protected] (Y.K.K.) These authors contributed equally to this work. y Received: 1 August 2019; Accepted: 29 August 2019; Published: 1 September 2019 Abstract: Epigenetic remodeling via histone acetylation has become a popular therapeutic strategy to treat Alzheimer’s disease (AD). In particular, histone deacetylase (HDAC) inhibitors including M344 and SAHA have been elucidated to be new drug candidates for AD, improving cognitive abilities impaired in AD mouse models. Although emerged as a promising target for AD, most of the HDAC inhibitors are poorly selective and could cause unwanted side effects. Here we show that tau is one of the cytosolic substrates of HDAC and the treatment of HDAC inhibitors such as Scriptaid, M344, BML281, and SAHA could increase the level of acetylated tau, resulting in the activation of tau pathology. Keywords: histone deacetylase inhibitor; tau acetylation; tau aggregation; Alzheimer’s disease 1. Introduction Alzheimer’s disease (AD) is a chronic neurodegenerative disorder that characterized by extracellular deposits of amyloid plaques and neuronal deposits of tau aggregates composed of hyperphosphorylated tau [1,2]. Over the last few decades, a number of compounds, designed to reduce the formation of amyloid plaques or to enhance their clearance, have failed in clinical trials. Since the causes of AD are still unknown, diverse targets are being applied for anti-Alzheimer’s drug discovery. Among the diverse, epigenetic regulation has been proposed to be a new promising therapeutic strategy for neurological disorders, particularly for AD [3]. In aged animal models, decreased levels of histone acetylation have been observed in the hippocampus and cerebral cortex [4,5]. Such changes could contribute to the development of neurodegeneration by down-regulating genes, which are critical for learning and memory. A number of recent studies have showed that histone deacetylase (HDAC) inhibitors exhibit neuro-protective properties, rescuing learning and memory abilities impaired in AD animal models [4,6–11]. Accordingly, HDAC inhibition has emerged as an alternative therapeutic strategy in AD treatment. Histone deacetylases are divided into four classes. Class I, II, and IV contain the classic HDAC enzymes, and Class III contains the sirtuin enzymes, which require NAD+ as a cofactor [12,13]. In 2009, Francis et al. proposed that epigenetic alteration by HDAC inhibition could be a therapeutic target to prevent AD progression [7]. In their study, trichostatin A, a HDAC inhibitor, rescued fear memory Int. J. Mol. Sci. 2019, 20, 4283; doi:10.3390/ijms20174283 www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2019, 20, 4283 2 of 12 impaired in APP/PS1 mice by increasing the acetylation of histone H4. In 2010, Peleg et al. reported that SAHA, a HDAC inhibitor, rescued age-dependent memory impairment in old mice by increasing the acetylation of histone H4 [4]. In 2017, Volmar et al. also suggested M344, a HDAC inhibitor, to be a promising drug candidate for AD [6]. In their study, M344 prevented cognitive declines in an AD animal model by down-regulating AD-related genes that contribute to APP processing and tau phosphorylation. Accumulating studies have supported that other HDAC inhibitors including valproic acid, 4-phenylbutyrate, MPT0G211, and nicotinamide presented similar therapeutic effects in AD animal models [8,10,14–16]. However, histone is not the only substrate of HDACs. A variety of non-histone substrates of HDACs exist in nuclei and cytosols [17,18]. Most HDAC inhibitors listed above are poorly selective and could cause unwanted side effects by acetylating non-histone proteins. In fact, several studies have showed that tau is a direct substrate of HDAC. In 2011, Cohen et al. reported that trichostatin A, a pan-HDAC inhibitor, increased tau acetylation [19]. In 2014, Noack et al. also reported that tubastatin A, a HDAC6 inhibitor, increased acetylated tau [20]. Tau is a neuron-specific microtubule-binding protein that stabilizes microtubules [21–23]. When pathologically modified, tau dissociates from microtubules and becomes insoluble aggregates [24–28]. Although hyperphosphorylation has been considered to be the major modification of tau, initiating tau pathology [2,29], recent studies have demonstrated that tau acetylation is also strongly associated with tau pathology [19,30,31]. Elevated levels of acetylated tau have been observed in AD patients [19,30], and acetylated tau has been colocalized with insoluble tau aggregates in the brain of AD animal models [19,32,33]. Biochemical studies have identified that tau acetylation slows down tau turnover, inhibiting proteasomal degradation [20,34]. In a result, acetylated tau accumulates, activating tau aggregation [19,30,34,35] and increased acetylated tau levels in the brain of tau transgenic mouse models are enough to cause an acetylation-mediated tau pathological cascade [32,33]. Due to the pathological implication, tau acetylation should be carefully evaluated in HDAC-targeting drug discovery. In this study, we collected 34 commercially available inhibitors of histone deacetylases (HDACs, SIRTs) and histone acetyl transferases (HATs) and evaluated the effect on tau acetylation and aggregation. Among the tested, pan-HDAC inhibitors (Scriptaid, M344, BML281, and SAHA) markedly induced intracellular tau aggregation. All the selected HDAC inhibitors increased the acetylation of tau at the residue K280 strongly as well as its representative cytosolic and nucleic substrates, tubulin and histone. 2. Results 2.1. Evaluation of HDAC Modulators on Tau-BiFC Sensor For the comprehensive evaluation of HDAC inhibitors, we collected 30 commercially available modulators of histone deacetylases (HDACs, SIRTs), together with 4 modulators of histone acetyl transferases (HATs). Then, the compound effects on tau aggregation were evaluated by using a tau aggregation sensor, named tau-BiFC [36]. As a fluorescence turn-on sensor, tau-BiFC fluorescence turns on only when tau assembles together (Figure1A). Tau-BiFC fluorescence directly represents the level of tau assembly in a cell, from soluble dimers to insoluble aggregates [36–40]. Forskolin was used as a positive control [36,41]. Among tested, 6 HDAC inhibitors, Scriptaid, M344, BML281, SAHA, Trichostatin A, and Fluoro-SAHA, induced tau-BiFC fluorescence noticeably by showing more than 3-fold increase at 3 µM concentration (Figure1B). In particular, Scriptaid, M344, BML281, and SAHA, HDAC inhibitors containing an aliphatic hydroxyamide linker acid, showed the strongest tau-BiFC fluorescence responses (Figures1C and2A). Int. J. Mol. Sci. 2019 , 20 , x FOR PEER REVIEW 3 of 12 evaluate acetylation levels of α-tubulin, a cytoplasmic substrate of HDACs, and histone H3, a nuclear substrate of HDACs [42,43]. The Tau-BiFC High group strikingly elevated both α-tubulin acetylation and histone H3 acetylation. The acetylation levels of α-tubulin increased over 3.0- up to 3.3-fold, and the acetylation levels of histone H3 increased over 3.5- up to 4.3-fold. In comparison, Tau-BiFC Null and Tau-BiFC Low groups did not show noticeable changes in α-tubulin acetylation (Figure 1D,E). In the Tau-BiFC Null group, BML210 and PhenylbutyrateNa slightly increased histone acetylation by showing 2.5- and 2.3-fold increases. The results indicate that Scriptaid, M344, BML281, and SAHA are pan-HDAC inhibitors, which strongly inhibit both cytoplasmic and nuclear HDACs. As a cytosolic substrate of HDACs, tau was also strongly acetylated by pan-HDAC inhibitors. Similar to Int. J.the Mol. increased Sci. 2019, 20 level, 4283 of acetylated tubulin, Tau(K280) acetylation increased almost 3-fold by the 3 of 12 treatment of the pan-HDAC inhibitors. Different from acetylated tubulin, acetylated tau seems accumulated in the cells, increasing the amount of total tau. FigureFigure 1. Evaluation 1. Evaluation of of HDAC HDAC modulators modulators on tau-BiFC tau-BiFC sensor. sensor. (A ()A Illustration) Illustration of tau-BiFC of tau-BiFC cell system. cell system. Tau-BiFCTau-BiFC cell expressescell expresses full-length full-length tau conjugatedtau conjugated with with the non-fluorescent the non-fluorescent N- or N- C-terminal or C-terminal fragments fragments of Venus fluorescence protein. When tau assembles together, Venus fluorescence turns on. of Venus fluorescence protein. When tau assembles together, Venus fluorescence turns on. (B) Screening (B) Screening of 34 compounds in tau-BiFC cells. Tau-BiFC cells were treated with 34 library of 34 compounds in tau-BiFC cells. Tau-BiFC cells were treated with 34 library compounds at 3 µM for 48 h. BiFC fluorescence intensities were quantified
Recommended publications
  • Histone Deacetylase Inhibitors: an Attractive Therapeutic Strategy Against Breast Cancer
    ANTICANCER RESEARCH 37 : 35-46 (2017) doi:10.21873/anticanres.11286 Review Histone Deacetylase Inhibitors: An Attractive Therapeutic Strategy Against Breast Cancer CHRISTOS DAMASKOS 1,2* , SERENA VALSAMI 3* , MICHAEL KONTOS 4* , ELEFTHERIOS SPARTALIS 2, THEODOROS KALAMPOKAS 5, EMMANOUIL KALAMPOKAS 6, ANTONIOS ATHANASIOU 4, DEMETRIOS MORIS 7, AFRODITE DASKALOPOULOU 2,8 , SPYRIDON DAVAKIS 4, GERASIMOS TSOUROUFLIS 1, KONSTANTINOS KONTZOGLOU 1, DESPINA PERREA 2, NIKOLAOS NIKITEAS 2 and DIMITRIOS DIMITROULIS 1 1Second Department of Propedeutic Surgery, 4First Department of Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 2N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 3Blood Transfusion Department, Aretaieion Hospital, Medical School, National and Kapodistrian Athens University, Athens, Greece; 5Assisted Conception Unit, Second Department of Obstetrics and Gynecology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; 6Gynaecological Oncology Department, University of Aberdeen, Aberdeen, U.K.; 7Lerner Research Institute, Cleveland Clinic, Cleveland, OH, U.S.A; 8School of Biology, National and Kapodistrian University of Athens, Athens, Greece Abstract. With a lifetime risk estimated to be one in eight in anticipate further clinical benefits of this new class of drugs, industrialized countries, breast cancer is the most frequent
    [Show full text]
  • Epigenetic Reprogramming of Tumor-Associated Fibroblasts in Lung Cancer: Therapeutic Opportunities
    cancers Review Epigenetic Reprogramming of Tumor-Associated Fibroblasts in Lung Cancer: Therapeutic Opportunities Jordi Alcaraz 1,2,3,*, Rafael Ikemori 1 , Alejandro Llorente 1 , Natalia Díaz-Valdivia 1 , Noemí Reguart 2,4 and Miguel Vizoso 5,* 1 Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain; [email protected] (R.I.); [email protected] (A.L.); [email protected] (N.D.-V.) 2 Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain; [email protected] 3 Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain 4 Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain 5 Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands * Correspondence: [email protected] (J.A.); [email protected] (M.V.) Simple Summary: Lung cancer is the leading cause of cancer death among both men and women, partly due to limited therapy responses. New avenues of knowledge are indicating that lung cancer cells do not form a tumor in isolation but rather obtain essential support from their surrounding host tissue rich in altered fibroblasts. Notably, there is growing evidence that tumor progression and even the current limited responses to therapies could be prevented by rescuing the normal behavior of fibroblasts, which are critical housekeepers of normal tissue function. For this purpose, it is key Citation: Alcaraz, J.; Ikemori, R.; to improve our understanding of the molecular mechanisms driving the pathologic alterations of Llorente, A.; Díaz-Valdivia, N.; fibroblasts in cancer.
    [Show full text]
  • Mechanisms and Clinical Significance of Histone Deacetylase Inhibitors: Epigenetic Glioblastoma Therapy
    ANTICANCER RESEARCH 35: 615-626 (2015) Review Mechanisms and Clinical Significance of Histone Deacetylase Inhibitors: Epigenetic Glioblastoma Therapy PHILIP LEE1*, BEN MURPHY1*, RICKEY MILLER1*, VIVEK MENON1*, NAREN L. BANIK1,2, PIERRE GIGLIO1,3, SCOTT M. LINDHORST1, ABHAY K. VARMA1, WILLIAM A. VANDERGRIFT III1, SUNIL J. PATEL1 and ARABINDA DAS1 1Department of Neurology and Neurosurgery & MUSC Brain & Spine Tumor Program Medical University of South Carolina, Charleston, SC, U.S.A.; 2Ralph H. Johnson VA Medical Center, Charleston, SC, U.S.A.; 3Department of Neurological Surgery Ohio State University Wexner Medical College, Columbus, OH, U.S.A. Abstract. Glioblastoma is the most common and deadliest glioblastoma therapy, explain the mechanisms of therapeutic of malignant primary brain tumors (Grade IV astrocytoma) effects as demonstrated by pre-clinical and clinical studies in adults. Current standard treatments have been improving and describe the current status of development of these drugs but patient prognosis still remains unacceptably devastating. as they pertain to glioblastoma therapy. Glioblastoma recurrence is linked to epigenetic mechanisms and cellular pathways. Thus, greater knowledge of the Glioblastoma (GBM) is the most common malignant adult cellular, genetic and epigenetic origin of glioblastoma is the brain tumor. Standard-of-care treatment includes surgery, key for advancing glioblastoma treatment. One rapidly radiation and temozolomide; however, this still yields poor growing field of treatment, epigenetic modifiers; histone prognosis for patients (1). Targeting of key epigenetic deacetylase inhibitors (HDACis), has now shown much enzymes, oncogenes and pathways specific to glioblastoma promise for improving patient outcomes through regulation cells by the drugs is very challenging, which has therefore of the acetylation states of histone proteins (a form of resulted in low potency in clinical trials (2).
    [Show full text]
  • Direct Amidation to Access 3-Amido-1,8-Naphthalimides Including Fluorescent Scriptaid Analogues As HDAC Inhibitors
    cells Article Direct Amidation to Access 3-Amido-1,8-Naphthalimides Including Fluorescent Scriptaid Analogues as HDAC Inhibitors Kyle N. Hearn 1,2, Trent D. Ashton 1,3,4 , Rameshwor Acharya 5, Zikai Feng 5 , Nuri Gueven 5 and Frederick M. Pfeffer 1,* 1 School of Life and Environmental Sciences, Deakin University, Waurn Ponds, VIC 3216, Australia 2 STEM College, RMIT University, Melbourne, VIC 3000, Australia; [email protected] 3 Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; [email protected] 4 Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia 5 School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7001, Australia; [email protected] (R.A.); [email protected] (Z.F.); [email protected] (N.G.) * Correspondence: [email protected] Abstract: Methodology to access fluorescent 3-amido-1,8-naphthalimides using direct Buchwald– Hartwig amidation is described. The protocol was successfully used to couple a number of substrates (including an alkylamide, an arylamide, a lactam and a carbamate) to 3-bromo-1,8-naphthalimide in good yield. To further exemplify the approach, a set of scriptaid analogues with amide substituents at the 3-position were prepared. The new compounds were more potent than scriptaid at a number of histone deacetylase (HDAC) isoforms including HDAC6. Activity was further confirmed in a whole cell tubulin deacetylation assay where the inhibitors were more active than the established Citation: Hearn, K.N.; Ashton, T.D.; HDAC6 selective inhibitor Tubastatin.
    [Show full text]
  • Histone Deacetylase Inhibitors for the Treatment of Myelodysplastic Syndrome and Acute Myeloid Leukemia
    Leukemia (2011) 25, 226–235 & 2011 Macmillan Publishers Limited All rights reserved 0887-6924/11 www.nature.com/leu REVIEW Histone deacetylase inhibitors for the treatment of myelodysplastic syndrome and acute myeloid leukemia A Quinta´s-Cardama1,3, FPS Santos1,2,3 and G Garcia-Manero1 1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA and 2Hematology Department, Hospital Israelita Albert Einstein, Sa˜o Paulo, Brazil Epigenetic changes have been identified in recent years as drugs whose mechanism of action has been linked to modula- important factors in the pathogenesis of myelodysplastic tion of gene transcription have taken center stage of both syndrome (MDS) and acute myeloid leukemia (AML). Histone clinical and preclinical research in AML and MDS.8 deacetylase inhibitors (HDACIs) regulate the acetylation of histones as well as other non-histone protein targets. Treat- Histone deacetylase inhibitors (HDACIs) represent one class ment with HDACIs results in chromatin remodeling that permits of gene expression modulating drugs, which is currently being re-expression of silenced tumor suppressor genes in cancer developed for therapy of several types of cancer.9 HDACIs were cells, which, in turn, can potentially result in cellular differ- initially developed as putative epigenetic agents, owing to their entiation, inhibition of proliferation and/or apoptosis. Several ability to modify acetylation status of histones and induce gene classes of HDACIs are currently under development for the expression.10 Typically, optimal gene expression occurs when treatment of patients with MDS and AML. Although modest 11 clinical activity has been reported with the use of HDACIs as histones are in a state of maximal acetylation.
    [Show full text]
  • China Japan United States Europe
    Inhibitor United States China Toll Free: +1 877 796-6397 Toll Free: 400-668-6834 Fax: +1 832 582-8590 Fax: 021-68591981 E-mail: [email protected] E-mail: [email protected] Europe Japan Tel: +49-89-46148500 Tel: + 81 3-5632-9610 Fax: +49-89-461485022 Fax: + 81 3-5632-9619 E-mail: [email protected] E-mail: [email protected] Protein Tyrosine Kinase PI3K/Akt/mTOR MAPK Inhibitors Cytoskeletal Signaling Selleck Chemicals supplies over 3,000 Apoptosis + inhibitors used in the study of cell 3000 Epigenetics Cell Cycle Inhibitors signaling pathways. Ubiquitin Signaling DNA Damage Stem Others Cells Neuronal Signaling Product Citations Selleck products have been cited in more than 12000 studies from various SCI journals. (Cell, Nature, Science: 61 studies) Compound Libraries Bioactive Compound Library 2659 compounds Nature. 2017, 541(7638):481-487. Nature. 2015, 521(7552):357-61. Science. 2016, 354(6315). Nature. 2017, 10.1038/nature21064. Nature. 2015, 521(7552):316-21. Science. 2016, 353(6302):929-32. Kinase Inhibitor Library Nature. 2016, 540(7631):119-123. Nature. 2015, 520(7549):683-7. Science. 2016, 352(6283):353-8. 430 inhibitors Nature. 2016, 539(7629):437-442. Nature. 2015, 520(7547):368-72. Science. 2016, 352(6282):189-96. Nature. 2016, 539(7628):304-308. Nature. 2015, 519(7543):370-3. Science. 2016, 351(6277):aad3680. FDA-approved Drug Library 1443 compounds Nature. 2016, 539(7627):54-58. Nature. 2015, 518(7538):254-7. Science. 2013, 341(6146):651-4. Nature. 2016, 538(7626):477-482. Nature. 2015, 517(7536):583-8.
    [Show full text]
  • Characterizing Heterogeneous Cellular Responses to Perturbations
    Characterizing heterogeneous cellular responses to perturbations Michael D. Slacka,b, Elisabeth D. Martineza,c, Lani F. Wua,b,1, and Steven J. Altschulera,b,1 aDepartment of Pharmacology, bGreen Center for Systems Biology, Simmons Cancer Center, and the cHamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390 Edited by Alfred G. Gilman, University of Texas Southwestern Medical Center, Dallas, TX, and approved October 16, 2008 (received for review July 20, 2008) Cellular populations have been widely observed to respond hetero- rithms to identify phenotypic ‘‘stereotypes’’ within the overall geneously to perturbation. However, interpreting the observed het- population and assign probabilities to cells belonging to subpopu- erogeneity is an extremely challenging problem because of the lations modeled on these stereotypes. Subpopulation identification complexity of possible cellular phenotypes, the large dimension of is based on application of a Gaussian mixture model (GMM), which potential perturbations, and the lack of methods for separating has been applied recently in the context of online phenotype meaningful biological information from noise. Here, we develop an discovery (30). Finally, for each population (or condition), we image-based approach to characterize cellular phenotypes based on estimated the fraction of cells in each subpopulation and summa- patterns of signaling marker colocalization. Heterogeneous cellular rized the results as a probability vector. The resulting ‘‘subpopula- populations are characterized as mixtures of phenotypically distinct tion profiles’’ are human- and machine-interpretable and enable the subpopulations, and responses to perturbations are summarized comparison of heterogeneous responses across multiple physiolog- succinctly as probabilistic redistributions of these mixtures. We apply ical, pathological, or environmental perturbations.
    [Show full text]
  • S13229-020-00387-6.Pdf
    Cavallo et al. Molecular Autism (2020) 11:88 https://doi.org/10.1186/s13229-020-00387-6 RESEARCH Open Access High-throughput screening identifes histone deacetylase inhibitors that modulate GTF2I expression in 7q11.23 microduplication autism spectrum disorder patient-derived cortical neurons Francesca Cavallo1†, Flavia Troglio2†, Giovanni Fagà3,4, Daniele Fancelli3,4, Reinald Shyti2, Sebastiano Trattaro1,2, Matteo Zanella2,10, Giuseppe D’Agostino2,11, James M. Hughes2,12, Maria Rosaria Cera3,4, Maurizio Pasi3,4, Michele Gabriele2,13, Maddalena Lazzarin1, Marija Mihailovich1,2, Frank Kooy9, Alessandro Rosa5,6, Ciro Mercurio3,4, Mario Varasi3,4 and Giuseppe Testa2,7,8* Abstract Background: Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental condition afecting almost 1% of children, and represents a major unmet medical need with no efective drug treatment available. Duplication at 7q11.23 (7Dup), encompassing 26–28 genes, is one of the best characterized ASD-causing copy number variations and ofers unique translational opportunities, because the hemideletion of the same interval causes Williams–Beuren syndrome (WBS), a condition defned by hypersociability and language strengths, thereby providing a unique refer- ence to validate treatments for the ASD symptoms. In the above-indicated interval at 7q11.23, defned as WBS critical region, several genes, such as GTF2I, BAZ1B, CLIP2 and EIF4H, emerged as critical for their role in the pathogenesis of WBS and 7Dup both from mouse models and human studies. Methods: We performed a high-throughput screening of 1478 compounds, including central nervous system agents, epigenetic modulators and experimental substances, on patient-derived cortical glutamatergic neurons dif- ferentiated from our cohort of induced pluripotent stem cell lines (iPSCs), monitoring the transcriptional modulation of WBS interval genes, with a special focus on GTF2I, in light of its overriding pathogenic role.
    [Show full text]
  • Histone Deacetylase Inhibitor Scriptaid Reactivates Latent HIV-1 Promoter by Inducing Histone Modification in in Vitro Latency Cell Lines
    265-272.qxd 18/6/2010 08:23 Ì ™ÂÏ›‰·265 INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 26: 265-272, 2010 265 Histone deacetylase inhibitor Scriptaid reactivates latent HIV-1 promoter by inducing histone modification in in vitro latency cell lines HAO YING, YUHAO ZHANG, SHIGUAN LIN, YEFEI HAN and HUAN-ZHANG ZHU State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China Received March 2, 2010; Accepted April 13, 2010 DOI: 10.3892/ijmm_00000461 Abstract. Human immunodeficiency virus type 1 (HIV-1) completely eradicate HIV-1 (1,2). It is now widely believed latency remains a major problem for the eradication of viruses that the presence of a long-lived, stable population of latently in infected individuals undergoing highly active anti-retroviral infected resting memory CD4+ T cells is a major barrier to therapy. By inhibiting HIV-1 gene expression and virus virus eradication (3-5). These latently infected cells can go production, histone deacetylase (HDAC) may contribute to the dormant, and remain in the peripheral circulation and other quiescence of HIV-1 within resting CD4+ T cells. A novel tissues for years despite effective HAART (5). The decay rate HDAC inhibitor, Scriptaid, has been found to have robust of the HIV-1 latent reservoir has been found to be so low that activity and lower toxicity compared to trichostatin A (TSA). their eradication during a human lifespan is unlikely (6,7). We therefore investigated Scriptaid for its capability to reverse Therefore, any curative AIDS therapy should provide a solution HIV-1 latency by inducing HIV-1 activation in the Jurkat T to the HIV-1 latency hurdle (3,6).
    [Show full text]
  • Anti-Tumor Activity of Phenoxybenzamine and Its Inhibition of Histone Deacetylases
    Touro Scholar NYMC Faculty Publications Faculty 6-13-2018 Anti-Tumor Activity of Phenoxybenzamine and Its Inhibition of Histone Deacetylases Mario A. Inchiosa New York Medical College Follow this and additional works at: https://touroscholar.touro.edu/nymc_fac_pubs Part of the Enzymes and Coenzymes Commons, and the Pharmacology Commons Recommended Citation Inchiosa, M. A. (2018). Anti-Tumor Activity of Phenoxybenzamine and Its Inhibition of Histone Deacetylases. PLoS One, 13 (6), 0198514-0198514. https://doi.org/10.1371/journal.pone.0198514 This Article is brought to you for free and open access by the Faculty at Touro Scholar. It has been accepted for inclusion in NYMC Faculty Publications by an authorized administrator of Touro Scholar. For more information, please contact [email protected]. RESEARCH ARTICLE Anti-tumor activity of phenoxybenzamine and its inhibition of histone deacetylases Mario A. Inchiosa, Jr.* Departments of Pharmacology and Anesthesiology, New York Medical College, Valhalla, New York, United States of America * [email protected] a1111111111 a1111111111 Abstract a1111111111 a1111111111 The principal finding from this study was the recognition that the α-adrenergic antagonist, a1111111111 phenoxybenzamine, possesses histone deacetylase inhibitory activity. Phenoxybenzamine is approved by the United States Food and Drug Administration for the treatment of hyper- tensive crises associated with tumors of the adrenal medulla, pheochromocytomas. It has several ªoff labelº indications relative to its capacity to relax vascular smooth muscle and OPEN ACCESS smooth muscle of the urogenital tract. The drug also has a long history of apparent efficacy Citation: Inchiosa MA, Jr. (2018) Anti-tumor in ameliorating, and perhaps reversing, the severe symptoms of neuropathic pain syn- activity of phenoxybenzamine and its inhibition of dromes.
    [Show full text]
  • A Database for Histone Deacetylase Inhibitors
    Journal name: Drug Design, Development and Therapy Article Designation: Methodology Year: 2015 Volume: 9 Drug Design, Development and Therapy Dovepress Running head verso: Murugan et al Running head recto: Database for histone deacetylase inhibitors open access to scientific and medical research DOI: http://dx.doi.org/10.2147/DDDT.S78276 Open Access Full Text Article METHODOLOGY HDACiDB: a database for histone deacetylase inhibitors Kasi Murugan1 Abstract: An histone deacetylase (HDAC) inhibitor database (HDACiDB) was constructed Shanmugasamy Sangeetha2 to enable rapid access to data relevant to the development of epigenetic modulators (HDAC Shanmugasamy Ranjitha2 inhibitors [HDACi]), helping bring precision cancer medicine a step closer. Thousands of Antony Vimala2 HDACi targeting HDACs are in various stages of development and are being tested in clinical Saleh Al-Sohaibani1 trials as monotherapy and in combination with other cancer agents. Despite the abundance of Gopal Rameshkumar2 HDACi, information resources are limited. Tools for in silico experiments on specific HDACi prediction, for designing and analyzing the generated data, as well as custom-made specific tools 1 Department of Botany and and interactive databases, are needed. We have developed an HDACiDB that is a composite Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia; collection of HDACi and currently comprises 1,445 chemical compounds, including 419 natural 2Bioinformatics Laboratory, Anna and 1,026 synthetic ones having the potential to inhibit histone deacetylation. Most importantly, University K. Balachander Research it will allow application of Lipinski’s rule of five drug-likeness and other physicochemical Centre, MIT Campus of Anna University Chennai, Chennai, India property-based screening of the inhibitors.
    [Show full text]
  • Uncovering Active Modulators of Native Macroautophagy Through
    bioRxiv preprint doi: https://doi.org/10.1101/756973; this version posted September 4, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 1 Title: Uncovering active modulators of native macroautophagy through novel 2 high-content screens 3 4 Author list: Safren N1, Tank EM1, Santoro N2, and Barmada SJ1* 5 6 Affiliations: 1University of Michigan, Department of Neurology, Ann Arbor MI, 2University 7 of Michigan, Center for Chemical Genomics, Life Sciences Institute 8 9 *Correspondence addressed to: [email protected] 10 11 12 Abstract 13 Autophagy is an evolutionarily conserved pathway mediating the breakdown of cellular 14 proteins and organelles. Emphasizing its pivotal nature, autophagy dysfunction 15 contributes to many diseases; nevertheless, development of effective autophagy 16 modulating drugs is hampered by fundamental deficiencies in available methods for 17 measuring autophagic activity, or flux. To overcome these limitations, we introduced the 18 photoconvertible protein Dendra2 into the MAP1LC3B locus of human cells via 19 CRISPR/Cas9 genome editing, enabling accurate and sensitive assessments of 20 autophagy in living cells by optical pulse labeling. High-content screening of 1,500 tool 21 compounds provided construct validity for the assay and uncovered many new 22 autophagy modulators. In an expanded screen of 24,000 diverse compounds, we 23 identified additional hits with profound effects on autophagy. Further, the autophagy 24 activator NVP-BEZ235 exhibited significant neuroprotective properties in a 25 neurodegenerative disease model. These studies confirm the utility of the Dendra2-LC3 26 assay, while simultaneously highlighting new autophagy-modulating compounds that 27 display promising therapeutic effects.
    [Show full text]